General Information of Drug Off-Target (DOT) (ID: OTRZX45T)

DOT Name Serine/threonine-protein kinase PLK1 (PLK1)
Synonyms EC 2.7.11.21; Polo-like kinase 1; PLK-1; Serine/threonine-protein kinase 13; STPK13
Gene Name PLK1
UniProt ID
PLK1_HUMAN
3D Structure
Download
2D Sequence (FASTA)
Download
3D Structure (PDB)
Download
PDB ID
1Q4K ; 1Q4O ; 1UMW ; 2OGQ ; 2OJX ; 2OU7 ; 2OWB ; 2RKU ; 2V5Q ; 2YAC ; 3BZI ; 3C5L ; 3FC2 ; 3FVH ; 3HIH ; 3HIK ; 3KB7 ; 3P2W ; 3P2Z ; 3P34 ; 3P35 ; 3P36 ; 3P37 ; 3Q1I ; 3RQ7 ; 3THB ; 4A4L ; 4A4O ; 4DFW ; 4E67 ; 4E9C ; 4E9D ; 4H5X ; 4H71 ; 4HAB ; 4HCO ; 4HY2 ; 4J52 ; 4J53 ; 4LKL ; 4LKM ; 4O56 ; 4O6W ; 4O9W ; 4RCP ; 4WHH ; 4WHK ; 4WHL ; 4X9R ; 4X9V ; 4X9W ; 5J19 ; 5NEI ; 5NFU ; 5NJE ; 5NMM ; 5NN1 ; 5NN2 ; 5TA6 ; 5TA8 ; 6AX4 ; 6GY2 ; 7MSO ; 7MX1 ; 8BJT ; 8CRC ; 8JOQ ; 8JOY
EC Number
2.7.11.21
Pfam ID
PF00069 ; PF00659
Sequence
MSAAVTAGKLARAPADPGKAGVPGVAAPGAPAAAPPAKEIPEVLVDPRSRRRYVRGRFLG
KGGFAKCFEISDADTKEVFAGKIVPKSLLLKPHQREKMSMEISIHRSLAHQHVVGFHGFF
EDNDFVFVVLELCRRRSLLELHKRRKALTEPEARYYLRQIVLGCQYLHRNRVIHRDLKLG
NLFLNEDLEVKIGDFGLATKVEYDGERKKTLCGTPNYIAPEVLSKKGHSFEVDVWSIGCI
MYTLLVGKPPFETSCLKETYLRIKKNEYSIPKHINPVAASLIQKMLQTDPTARPTINELL
NDEFFTSGYIPARLPITCLTIPPRFSIAPSSLDPSNRKPLTVLNKGLENPLPERPREKEE
PVVRETGEVVDCHLSDMLQQLHSVNASKPSERGLVRQEEAEDPACIPIFWVSKWVDYSDK
YGLGYQLCDNSVGVLFNDSTRLILYNDGDSLQYIERDGTESYLTVSSHPNSLMKKITLLK
YFRNYMSEHLLKAGANITPREGDELARLPYLRTWFRTRSAIILHLSNGSVQINFFQDHTK
LILCPLMAAVTYIDEKRDFRTYRLSLLEEYGCCKELASRLRYARTMVDKLLSSRSASNRL
KAS
Function
Serine/threonine-protein kinase that performs several important functions throughout M phase of the cell cycle, including the regulation of centrosome maturation and spindle assembly, the removal of cohesins from chromosome arms, the inactivation of anaphase-promoting complex/cyclosome (APC/C) inhibitors, and the regulation of mitotic exit and cytokinesis. Polo-like kinase proteins act by binding and phosphorylating proteins that are already phosphorylated on a specific motif recognized by the POLO box domains. Phosphorylates BORA, BUB1B/BUBR1, CCNB1, CDC25C, CEP55, ECT2, ERCC6L, FBXO5/EMI1, FOXM1, KIF20A/MKLP2, CENPU, NEDD1, NINL, NPM1, NUDC, PKMYT1/MYT1, KIZ, PPP1R12A/MYPT1, POLQ, PRC1, RACGAP1/CYK4, RAD51, RHNO1, SGO1, STAG2/SA2, TEX14, TOPORS, p73/TP73, TPT1, WEE1 and HNRNPU. Plays a key role in centrosome functions and the assembly of bipolar spindles by phosphorylating KIZ, NEDD1 and NINL. NEDD1 phosphorylation promotes subsequent targeting of the gamma-tubulin ring complex (gTuRC) to the centrosome, an important step for spindle formation. Phosphorylation of NINL component of the centrosome leads to NINL dissociation from other centrosomal proteins. Involved in mitosis exit and cytokinesis by phosphorylating CEP55, ECT2, KIF20A/MKLP2, CENPU, PRC1 and RACGAP1. Recruited at the central spindle by phosphorylating and docking PRC1 and KIF20A/MKLP2; creates its own docking sites on PRC1 and KIF20A/MKLP2 by mediating phosphorylation of sites subsequently recognized by the POLO box domains. Phosphorylates RACGAP1, thereby creating a docking site for the Rho GTP exchange factor ECT2 that is essential for the cleavage furrow formation. Promotes the central spindle recruitment of ECT2. Plays a central role in G2/M transition of mitotic cell cycle by phosphorylating CCNB1, CDC25C, FOXM1, CENPU, PKMYT1/MYT1, PPP1R12A/MYPT1 and WEE1. Part of a regulatory circuit that promotes the activation of CDK1 by phosphorylating the positive regulator CDC25C and inhibiting the negative regulators WEE1 and PKMYT1/MYT1. Also acts by mediating phosphorylation of cyclin-B1 (CCNB1) on centrosomes in prophase. Phosphorylates FOXM1, a key mitotic transcription regulator, leading to enhance FOXM1 transcriptional activity. Involved in kinetochore functions and sister chromatid cohesion by phosphorylating BUB1B/BUBR1, FBXO5/EMI1 and STAG2/SA2. PLK1 is high on non-attached kinetochores suggesting a role of PLK1 in kinetochore attachment or in spindle assembly checkpoint (SAC) regulation. Required for kinetochore localization of BUB1B. Regulates the dissociation of cohesin from chromosomes by phosphorylating cohesin subunits such as STAG2/SA2. Phosphorylates SGO1: required for spindle pole localization of isoform 3 of SGO1 and plays a role in regulating its centriole cohesion function. Mediates phosphorylation of FBXO5/EMI1, a negative regulator of the APC/C complex during prophase, leading to FBXO5/EMI1 ubiquitination and degradation by the proteasome. Acts as a negative regulator of p53 family members: phosphorylates TOPORS, leading to inhibit the sumoylation of p53/TP53 and simultaneously enhance the ubiquitination and subsequent degradation of p53/TP53. Phosphorylates the transactivation domain of the transcription factor p73/TP73, leading to inhibit p73/TP73-mediated transcriptional activation and pro-apoptotic functions. Phosphorylates BORA, and thereby promotes the degradation of BORA. Contributes to the regulation of AURKA function. Also required for recovery after DNA damage checkpoint and entry into mitosis. Phosphorylates MISP, leading to stabilization of cortical and astral microtubule attachments required for proper spindle positioning. Together with MEIKIN, acts as a regulator of kinetochore function during meiosis I: required both for mono-orientation of kinetochores on sister chromosomes and protection of centromeric cohesin from separase-mediated cleavage. Phosphorylates CEP68 and is required for its degradation. Regulates nuclear envelope breakdown during prophase by phosphorylating DCTN1 resulting in its localization in the nuclear envelope. Phosphorylates the heat shock transcription factor HSF1, promoting HSF1 nuclear translocation upon heat shock. Phosphorylates HSF1 also in the early mitotic period; this phosphorylation regulates HSF1 localization to the spindle pole, the recruitment of the SCF(BTRC) ubiquitin ligase complex induicing HSF1 degradation, and hence mitotic progression. Regulates mitotic progression by phosphorylating RIOK2. Through the phosphorylation of DZIP1 regulates the localization during mitosis of the BBSome, a ciliary protein complex involved in cilium biogenesis. Regulates DNA repair during mitosis by mediating phosphorylation of POLQ and RHNO1, thereby promoting POLQ recruitment to DNA damage sites.
Tissue Specificity Placenta and colon.
KEGG Pathway
FoxO sig.ling pathway (hsa04068 )
Cell cycle (hsa04110 )
Oocyte meiosis (hsa04114 )
Progesterone-mediated oocyte maturation (hsa04914 )
Reactome Pathway
Polo-like kinase mediated events (R-HSA-156711 )
Golgi Cisternae Pericentriolar Stack Reorganization (R-HSA-162658 )
APC/C (R-HSA-174178 )
Phosphorylation of the APC/C (R-HSA-176412 )
Phosphorylation of Emi1 (R-HSA-176417 )
Condensation of Prophase Chromosomes (R-HSA-2299718 )
Separation of Sister Chromatids (R-HSA-2467813 )
Resolution of Sister Chromatid Cohesion (R-HSA-2500257 )
Regulation of PLK1 Activity at G2/M Transition (R-HSA-2565942 )
Activation of NIMA Kinases NEK9, NEK6, NEK7 (R-HSA-2980767 )
Loss of Nlp from mitotic centrosomes (R-HSA-380259 )
Recruitment of mitotic centrosome proteins and complexes (R-HSA-380270 )
Loss of proteins required for interphase microtubule organization from the centrosome (R-HSA-380284 )
Recruitment of NuMA to mitotic centrosomes (R-HSA-380320 )
Anchoring of the basal body to the plasma membrane (R-HSA-5620912 )
RHO GTPases Activate Formins (R-HSA-5663220 )
Mitotic Prometaphase (R-HSA-68877 )
Mitotic Metaphase/Anaphase Transition (R-HSA-68881 )
Mitotic Telophase/Cytokinesis (R-HSA-68884 )
Cyclin A/B1/B2 associated events during G2/M transition (R-HSA-69273 )
The role of GTSE1 in G2/M progression after G2 checkpoint (R-HSA-8852276 )
AURKA Activation by TPX2 (R-HSA-8854518 )
EML4 and NUDC in mitotic spindle formation (R-HSA-9648025 )
Amplification of signal from unattached kinetochores via a MAD2 inhibitory signal (R-HSA-141444 )

Molecular Interaction Atlas (MIA) of This DOT

Molecular Interaction Atlas (MIA) Jump to Detail Molecular Interaction Atlas of This DOT
This DOT Affected the Drug Response of 1 Drug(s)
Drug Name Drug ID Highest Status Interaction REF
Imatinib DM7RJXL Approved Serine/threonine-protein kinase PLK1 (PLK1) increases the response to substance of Imatinib. [45]
------------------------------------------------------------------------------------
44 Drug(s) Affected the Gene/Protein Processing of This DOT
Drug Name Drug ID Highest Status Interaction REF
Valproate DMCFE9I Approved Valproate decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [1]
Ciclosporin DMAZJFX Approved Ciclosporin decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [2]
Tretinoin DM49DUI Approved Tretinoin decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [3]
Doxorubicin DMVP5YE Approved Doxorubicin decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [4]
Cupric Sulfate DMP0NFQ Approved Cupric Sulfate decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [5]
Cisplatin DMRHGI9 Approved Cisplatin decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [6]
Estradiol DMUNTE3 Approved Estradiol increases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [7]
Ivermectin DMDBX5F Approved Ivermectin decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [8]
Quercetin DM3NC4M Approved Quercetin decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [9]
Hydrogen peroxide DM1NG5W Approved Hydrogen peroxide decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [11]
Calcitriol DM8ZVJ7 Approved Calcitriol decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [12]
Testosterone DM7HUNW Approved Testosterone decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [12]
Methotrexate DM2TEOL Approved Methotrexate decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [13]
Zoledronate DMIXC7G Approved Zoledronate decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [14]
Menadione DMSJDTY Approved Menadione decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [11]
Fluorouracil DMUM7HZ Approved Fluorouracil decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [4]
Cannabidiol DM0659E Approved Cannabidiol decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [15]
Troglitazone DM3VFPD Approved Troglitazone decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [16]
Irinotecan DMP6SC2 Approved Irinotecan decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [17]
Dasatinib DMJV2EK Approved Dasatinib decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [18]
Gemcitabine DMSE3I7 Approved Gemcitabine decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [19]
Lucanthone DMZLBUO Approved Lucanthone decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [20]
Palbociclib DMD7L94 Approved Palbociclib decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [21]
Resveratrol DM3RWXL Phase 3 Resveratrol decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [22]
Curcumin DMQPH29 Phase 3 Curcumin decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [23]
Genistein DM0JETC Phase 2/3 Genistein decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [24]
phorbol 12-myristate 13-acetate DMJWD62 Phase 2 phorbol 12-myristate 13-acetate increases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [25]
PEITC DMOMN31 Phase 2 PEITC decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [26]
R-roscovitine DMSH108 Phase 2 R-roscovitine decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [27]
(+)-JQ1 DM1CZSJ Phase 1 (+)-JQ1 decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [29]
Leflunomide DMR8ONJ Phase 1 Trial Leflunomide decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [30]
Mivebresib DMCPF90 Phase 1 Mivebresib decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [31]
TAK-114 DMTXE19 Phase 1 TAK-114 decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [32]
PMID28460551-Compound-2 DM4DOUB Patented PMID28460551-Compound-2 decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [33]
Flavonoid derivative 1 DMCQP0B Patented Flavonoid derivative 1 decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [35]
THAPSIGARGIN DMDMQIE Preclinical THAPSIGARGIN increases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [36]
Bisphenol A DM2ZLD7 Investigative Bisphenol A decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [37]
Trichostatin A DM9C8NX Investigative Trichostatin A affects the expression of Serine/threonine-protein kinase PLK1 (PLK1). [38]
Coumestrol DM40TBU Investigative Coumestrol increases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [39]
Sulforaphane DMQY3L0 Investigative Sulforaphane increases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [40]
Paraquat DMR8O3X Investigative Paraquat increases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [41]
geraniol DMS3CBD Investigative geraniol decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [42]
Dibutyl phthalate DMEDGKO Investigative Dibutyl phthalate increases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [32]
I-BET151 DMYRUH2 Investigative I-BET151 decreases the expression of Serine/threonine-protein kinase PLK1 (PLK1). [44]
------------------------------------------------------------------------------------
⏷ Show the Full List of 44 Drug(s)
6 Drug(s) Affected the Post-Translational Modifications of This DOT
Drug Name Drug ID Highest Status Interaction REF
Arsenic trioxide DM61TA4 Approved Arsenic trioxide increases the phosphorylation of Serine/threonine-protein kinase PLK1 (PLK1). [10]
Hydroxyurea DMOQVU9 Approved Hydroxyurea decreases the phosphorylation of Serine/threonine-protein kinase PLK1 (PLK1). [10]
Benzo(a)pyrene DMN7J43 Phase 1 Benzo(a)pyrene decreases the methylation of Serine/threonine-protein kinase PLK1 (PLK1). [28]
PMID28870136-Compound-52 DMFDERP Patented PMID28870136-Compound-52 decreases the phosphorylation of Serine/threonine-protein kinase PLK1 (PLK1). [34]
Coumarin DM0N8ZM Investigative Coumarin increases the phosphorylation of Serine/threonine-protein kinase PLK1 (PLK1). [34]
Microcystin-LR DMTMLRN Investigative Microcystin-LR increases the phosphorylation of Serine/threonine-protein kinase PLK1 (PLK1). [43]
------------------------------------------------------------------------------------
⏷ Show the Full List of 6 Drug(s)

References

1 A genomic approach to predict synergistic combinations for breast cancer treatment. Pharmacogenomics J. 2013 Feb;13(1):94-104. doi: 10.1038/tpj.2011.48. Epub 2011 Nov 15.
2 Comparison of HepG2 and HepaRG by whole-genome gene expression analysis for the purpose of chemical hazard identification. Toxicol Sci. 2010 May;115(1):66-79.
3 Effect of retinoic acid on gene expression in human conjunctival epithelium: secretory phospholipase A2 mediates retinoic acid induction of MUC16. Invest Ophthalmol Vis Sci. 2005 Nov;46(11):4050-61.
4 Cell-type-specific responses to chemotherapeutics in breast cancer. Cancer Res. 2004 Jun 15;64(12):4218-26.
5 Physiological and toxicological transcriptome changes in HepG2 cells exposed to copper. Physiol Genomics. 2009 Aug 7;38(3):386-401.
6 Low doses of cisplatin induce gene alterations, cell cycle arrest, and apoptosis in human promyelocytic leukemia cells. Biomark Insights. 2016 Aug 24;11:113-21.
7 FOXM1 is a transcriptional target of ERalpha and has a critical role in breast cancer endocrine sensitivity and resistance. Oncogene. 2010 May 20;29(20):2983-95. doi: 10.1038/onc.2010.47. Epub 2010 Mar 8.
8 Quantitative proteomics reveals a broad-spectrum antiviral property of ivermectin, benefiting for COVID-19 treatment. J Cell Physiol. 2021 Apr;236(4):2959-2975. doi: 10.1002/jcp.30055. Epub 2020 Sep 22.
9 Comparison of phenotypic and transcriptomic effects of false-positive genotoxins, true genotoxins and non-genotoxins using HepG2 cells. Mutagenesis. 2011 Sep;26(5):593-604.
10 PLK1 targets NOTCH1 during DNA damage and mitotic progression. J Biol Chem. 2019 Nov 22;294(47):17941-17950. doi: 10.1074/jbc.RA119.009881. Epub 2019 Oct 9.
11 Gene expression after treatment with hydrogen peroxide, menadione, or t-butyl hydroperoxide in breast cancer cells. Cancer Res. 2002 Nov 1;62(21):6246-54.
12 Effects of 1alpha,25 dihydroxyvitamin D3 and testosterone on miRNA and mRNA expression in LNCaP cells. Mol Cancer. 2011 May 18;10:58.
13 Methotrexate modulates folate phenotype and inflammatory profile in EA.hy 926 cells. Eur J Pharmacol. 2014 Jun 5;732:60-7.
14 The proapoptotic effect of zoledronic acid is independent of either the bone microenvironment or the intrinsic resistance to bortezomib of myeloma cells and is enhanced by the combination with arsenic trioxide. Exp Hematol. 2011 Jan;39(1):55-65.
15 Cannabidiol-induced transcriptomic changes and cellular senescence in human Sertoli cells. Toxicol Sci. 2023 Feb 17;191(2):227-238. doi: 10.1093/toxsci/kfac131.
16 Effects of ciglitazone and troglitazone on the proliferation of human stomach cancer cells. World J Gastroenterol. 2009 Jan 21;15(3):310-20.
17 In vitro and in vivo irinotecan-induced changes in expression profiles of cell cycle and apoptosis-associated genes in acute myeloid leukemia cells. Mol Cancer Ther. 2005 Jun;4(6):885-900.
18 Dasatinib reverses cancer-associated fibroblasts (CAFs) from primary lung carcinomas to a phenotype comparable to that of normal fibroblasts. Mol Cancer. 2010 Jun 27;9:168.
19 A fine-needle aspirate-based vulnerability assay identifies polo-like kinase 1 as a mediator of gemcitabine resistance in pancreatic cancer. Mol Cancer Ther. 2010 Feb;9(2):311-8. doi: 10.1158/1535-7163.MCT-09-0693. Epub 2010 Jan 26.
20 Lucanthone is a novel inhibitor of autophagy that induces cathepsin D-mediated apoptosis. J Biol Chem. 2011 Feb 25;286(8):6602-13.
21 Cdk4/6 inhibition induces epithelial-mesenchymal transition and enhances invasiveness in pancreatic cancer cells. Mol Cancer Ther. 2012 Oct;11(10):2138-48. doi: 10.1158/1535-7163.MCT-12-0562. Epub 2012 Aug 6.
22 Resveratrol-induced gene expression profiles in human prostate cancer cells. Cancer Epidemiol Biomarkers Prev. 2005 Mar;14(3):596-604. doi: 10.1158/1055-9965.EPI-04-0398.
23 Novel carbocyclic curcumin analog CUR3d modulates genes involved in multiple apoptosis pathways in human hepatocellular carcinoma cells. Chem Biol Interact. 2015 Dec 5;242:107-22.
24 Dose- and time-dependent transcriptional response of Ishikawa cells exposed to genistein. Toxicol Sci. 2016 May;151(1):71-87.
25 Comparison of gene expression profiles in HepG2 cells exposed to arsenic, cadmium, nickel, and three model carcinogens for investigating the mechanisms of metal carcinogenesis. Environ Mol Mutagen. 2009 Jan;50(1):46-59.
26 Phenethyl isothiocyanate alters the gene expression and the levels of protein associated with cell cycle regulation in human glioblastoma GBM 8401 cells. Environ Toxicol. 2017 Jan;32(1):176-187.
27 Proliferative suppression by CDK4/6 inhibition: complex function of the retinoblastoma pathway in liver tissue and hepatoma cells. Gastroenterology. 2010 May;138(5):1920-30. doi: 10.1053/j.gastro.2010.01.007. Epub 2010 Jan 25.
28 Air pollution and DNA methylation alterations in lung cancer: A systematic and comparative study. Oncotarget. 2017 Jan 3;8(1):1369-1391. doi: 10.18632/oncotarget.13622.
29 Inhibition of BRD4 attenuates tumor cell self-renewal and suppresses stem cell signaling in MYC driven medulloblastoma. Oncotarget. 2014 May 15;5(9):2355-71.
30 Endoplasmic reticulum stress and MAPK signaling pathway activation underlie leflunomide-induced toxicity in HepG2 Cells. Toxicology. 2017 Dec 1;392:11-21.
31 Superior efficacy of cotreatment with BET protein inhibitor and BCL2 or MCL1 inhibitor against AML blast progenitor cells. Blood Cancer J. 2019 Jan 15;9(2):4. doi: 10.1038/s41408-018-0165-5.
32 In silico, in vitro and in vivo studies: Dibutyl phthalate promotes prostate cancer cell proliferation by activating Forkhead Box M1 and remission after Natura- pretreatment. Toxicology. 2023 Apr;488:153465. doi: 10.1016/j.tox.2023.153465. Epub 2023 Feb 23.
33 Cell-based two-dimensional morphological assessment system to predict cancer drug-induced cardiotoxicity using human induced pluripotent stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol. 2019 Nov 15;383:114761. doi: 10.1016/j.taap.2019.114761. Epub 2019 Sep 15.
34 Quantitative phosphoproteomics reveal cellular responses from caffeine, coumarin and quercetin in treated HepG2 cells. Toxicol Appl Pharmacol. 2022 Aug 15;449:116110. doi: 10.1016/j.taap.2022.116110. Epub 2022 Jun 7.
35 Luteolin exerts anti-tumor activity through the suppression of epidermal growth factor receptor-mediated pathway in MDA-MB-231 ER-negative breast cancer cells. Food Chem Toxicol. 2012 Nov;50(11):4136-43. doi: 10.1016/j.fct.2012.08.025. Epub 2012 Aug 20.
36 The genome-wide expression profile of Scrophularia ningpoensis-treated thapsigargin-stimulated U-87MG cells. Neurotoxicology. 2009 May;30(3):368-76.
37 Bisphenol A induces DSB-ATM-p53 signaling leading to cell cycle arrest, senescence, autophagy, stress response, and estrogen release in human fetal lung fibroblasts. Arch Toxicol. 2018 Apr;92(4):1453-1469.
38 A trichostatin A expression signature identified by TempO-Seq targeted whole transcriptome profiling. PLoS One. 2017 May 25;12(5):e0178302. doi: 10.1371/journal.pone.0178302. eCollection 2017.
39 Pleiotropic combinatorial transcriptomes of human breast cancer cells exposed to mixtures of dietary phytoestrogens. Food Chem Toxicol. 2009 Apr;47(4):787-95.
40 Transcriptome and DNA methylation changes modulated by sulforaphane induce cell cycle arrest, apoptosis, DNA damage, and suppression of proliferation in human liver cancer cells. Food Chem Toxicol. 2020 Feb;136:111047. doi: 10.1016/j.fct.2019.111047. Epub 2019 Dec 12.
41 An in vitro strategy using multiple human induced pluripotent stem cell-derived models to assess the toxicity of chemicals: A case study on paraquat. Toxicol In Vitro. 2022 Jun;81:105333. doi: 10.1016/j.tiv.2022.105333. Epub 2022 Feb 16.
42 Geraniol suppresses prostate cancer growth through down-regulation of E2F8. Cancer Med. 2016 Oct;5(10):2899-2908.
43 Microcystin-LR promotes proliferation by activating Akt/S6K1 pathway and disordering apoptosis and cell cycle associated proteins phosphorylation in HL7702 cells. Toxicol Lett. 2016 Jan 5;240(1):214-25. doi: 10.1016/j.toxlet.2015.10.015. Epub 2015 Oct 23.
44 BET Bromodomain Inhibition as a Therapeutic Strategy in Ovarian Cancer by Downregulating FoxM1. Theranostics. 2016 Jan 1;6(2):219-30. doi: 10.7150/thno.13178. eCollection 2016.
45 Efficacy of the polo-like kinase inhibitor rigosertib, alone or in combination with Abelson tyrosine kinase inhibitors, against break point cluster region-c-Abelson-positive leukemia cells. Oncotarget. 2015 Aug 21;6(24):20231-40. doi: 10.18632/oncotarget.4047.